1 Background
Chimeric antigen receptor (CAR)-T cell therapy has revolutionized the immunotherapy field with outstanding results in the treatment of several relapsed/refractory (RR) hematological malignancies. CAR-T cell therapy directed against B-cell maturation antigen (BCMA) is under clinical investigation for the treatment of multiple myeloma (MM). In the KarMMa study, Idecabtagene vicleucel (idecel) achieved an overall response rate (ORR) of 73% and the median progression-free survival (PFS) was 8.8 months [
1]. Ciltacabtagene autoleucel (cilta-cel) showed an ORR of 97.9% at 27.7 months with a median PFS of 34.9 months in the CARTITUDE-1 study [
2‐
5]. However, although real-world evidence remains limited in BCMA-directed CAR-T therapy, idecel and cilta-cel have shown 1-year progression rates of 60% and 33%, respectively, in clinical trials [
1,
3,
6]. Therefore, relapse ultimately remains common with all approved anti-BCMA CAR-T products [
7]. In this sense, several limitations have been identified that may explain the ultimate relapse of patients. The heterogeneity of BCMA expression levels in patients, the presence of soluble BCMA in blood, on-/off-tumor toxicity, the lack of CAR-T cell persistence, and poor quality of T-cell products are some of the limitations that may affect CAR-T cell therapy in MM [
8]. The development of next-generation CAR-T cells is mandatory to overcome restrictions to CAR-T efficacy observed in the real-world setting.
We present a novel academic BCMA CAR-T product (CARTemis-1) developed following a rational design for enhanced antitumoral efficacy against plasma cells. Remarkably, CARTemis-1 is not hindered by soluble BCMA and contains a safety-gene marker. CARTemis-1 shows potent antitumor efficacy in preclinical models, both in vitro and in vivo, and its manufacture has been validated under GMP conditions. Importantly, little attention has been given to what happens during ex-vivo expansion and maintenance of CAR-T cells and the effect of the manufacturing process on the dynamics of the immunophenotype of cell products. In this study, CAR-T cells produced under GMP conditions were monitored during the manufacturing process, and immunophenotypic and functional characterization were performed to identify the optimal time for expansion. In addition, the dynamics of CAR-T cells during the manufacturing process, and how manufacturing process impacts CAR-T cell features and quality were assessed. A phase I/II clinical trial for patients with multiple myeloma will be conducted (EuCT number 2022-503063-15-00).
2 Methods
2.1 CAR gene construct and design
A codon-optimized single chain variable fragment (scFv) comprising the variable heavy (VH) and variable light (VL) chains of the anti-BCMA mAb BCMA30 [
9] (separated by two different (G4S)3 linkers, were synthesized (GeneArt, Regensburg, Germany) and fused to a different spacer and a transmembrane domain, a 4-1BB_CD3𝜁 signaling module, a T2A element and a safety-gene composed of a truncated epidermal-growth -factor-receptor (EGFRt) in an epHIV7 lentiviral vector backbone.
2.2 In vitro studies with CARTemis-1
Cytotoxic activity was analyzed in a bioluminescence-based assay using firefly luciferase (ffluc)-transduced target cells and distinct effector-to-target cell (E: T) ratios at 4 h post-coculture. Assay was performed with 5 × 10
3 target cells/well with effector T cells at various E: T ratios in triplicate wells. Specific lysis was calculated using a standard formula [
10]:
$$\eqalign{&\% ~specific\,lysis = \cr &\quad\quad\left({\frac{Mean\,bioluminiscence\,of\,target\,cells\,-\,Mean\,bioluminiscence\,of\,coculture}{Mean\,bioluminiscence\,of\,target\,cells~~}}\right) * 100} $$
Proliferation of T cells was analyzed by carboxyfluorescein succinimidyl ester (CFSE) (Biolegend, Ref: 423801) dye dilution after 72 h of coculture with target cells (E: T ratio of 4:1). IFN-γ, IL-2 and GM-CSF secretion in supernatants obtained after 24 h of coculture of T cells with target cells (E: T ratio of 4:1) were measured by enzyme-linked immunosorbant assay (ELISA) (Biolegend). Assay was performed in triplicate wells with 5 × 103 target cells per well. Recombinant soluble BCMA protein was obtained from R&D Systems (R&D Systems, Ref: ATM193). Primary myeloma cells were obtained from bone-marrow samples from patients with multiple myeloma at the time of diagnosis and were isolated using MACSprep™ Multiple Myeloma CD138 MicroBeads Human from Miltenyi Biotec (Ref: 130-111-744) and an AutoMACS Pro Separator (Miltenyi Biotec).
2.3 In vivo studies with CARTemis-1
The University of Würzburg Institutional Animal Care and Use Committee and the Institutional Animal Care and Use Committee at the Institute of Biomedicine in Seville approved all animal procedures. Six-to-eight-week-old NSG (NOD-scid IL2rγnull) mice were obtained from Charles River (Sulzfeld, Germany). Tumor cells, PBMCs, or CAR-T cells were inoculated via the tail injection vein, and animals were randomly allocated to the treatment groups. Specific methods used for the three different mouse models are described in Supplementary Material.
2.4 GMP-manufacture of CARTemis-1
Apheresis products from healthy donors were connected to CliniMACS Prodigy system (Miltenyi Biotec, Germany). CD4+/CD8+ white blood cells were selected using coated-magnetic beads (Miltenyi Biotec, Ref: 200-070-213, and Ref: 200-070-215, respectively). A total of 1 × 108 cells were used to initiate cell culture with TexMACs (Miltenyi Biotec, Ref: 170-076-306) supplemented with IL-7 and IL-15 (Miltenyi Biotec, Ref: 170-076-184, and Ref: 170-076-114, respectively). Cells were activated using TransACT (Miltenyi Biotec, Ref: 200-076-204) and transduced at a multiplicity of infection of 5 at day 2 post-activation. In vitro cytotoxicity, cytokines, and quality control assays were performed on days 8, 9, and 10 post activation. CARTemis-1 products were frozen according to dose and thawed to perform stability studies at 6 and 12 months after cryopreservation. A graphical summary of GMP-manufacture of CARTemis-1 in CliniMACS Prodigy is included in Supplementary Material. Additional information regarding quality controls according to GMP standards is described in Supplementary Material.
2.5 Statistical analysis
Statistical analyses were performed using Prism software v6.07 (GraphPad, San Diego, California). Unpaired and paired t-tests were used to analyze data from in vitro experiments. P values < 0.05 were considered to indicate statistical significance. Survival curves were analyzed using Log-rank (Mantel-cox) test. The measurements per group are presented as the means and standard deviations. When the qq plots showed no normally distributed data and due to the small sample size, nonparametric Mann-Whitney U tests were used as significance tests for comparing the measurements between groups.
4 Discussion
Despite the impressive results of CAR-T cell therapy in MM, approximately 45% of patients relapse at 24 months of treatment according to the CARTITUDE-4 trial [
5]. Several limitations have been identified and CAR construct optimization is under investigation. Here, we present the preclinical and clinical validation of a next-generation BCMA CAR-T cell (CARTemis-1).
The connecting sequence between the recognition domain to the transmembrane domain can profoundly affect CAR T-cell function by altering the length and flexibility of the resulting CAR [
11]. According to these findings, previous studies have shown that the length of the CAR construct should be adapted to the specific target antigen to optimize antigen binding [
11,
12]. In this regard, several studies have shown that the optimal spacer length depends on the distance of the targeted epitope from the tumor cell membrane [
13‐
15]. Our study provides another example of how CAR flexibility can be modulated with different extracellular spacers to increase antitumor efficacy. In our study, CARTemis-1 with a longer spacer region, showed increased production of antitumor cytokines and increased cytotoxicity against myeloma cells. This is in accordance with previous studies in which longer spacers provided flexibility to the CAR and allowed for better access to membrane-proximal epitopes [
11,
13,
14]. In this regard, CARTemis-1 recognizes a membrane-proximal epitope (24–41 AA) of the extracellular domain of BCMA. In addition, the extracellular domain of BCMA is very short (54 AA, UniProt: A7KBT6) compared to that of other proteins (CD19: 272 AA, UniProt: P15391; CD20: 668 AA, UniProt: P11836), suggesting that the long spacer of CARTemis-1 offers better flexibility to access the target.
In MM, BCMA is the preferred antigen due to its specific expression in plasma cells, with minimal expression in hematopoietic stem cells and normal tissues [
16,
17]. However, the BCMA extracellular domain can be shed by the protease γ-secretase, and a soluble form of BCMA protein is secreted into the blood. This fact is one of the limitations of current anti-BCMA CAR-T therapy and is related to the loss of CAR-T efficacy [
18‐
22]. In fact, some studies have shown increased levels of sBCMA in the serum of patients who do not respond to anti-BCMA CAR-T therapy [
23,
24]. Multiple studies have demonstrated how increased levels of soluble BCMA affect the antitumor efficacy of BCMA CAR-T cells [
18‐
20,
25]. Titration assays with recombinant soluble BCMA protein showed that CAR-T cell activity was inhibited at concentrations up to 32 ng/mL of BCMA [
18]. Additionally, sBCMA at just 10 ng/mL significantly reduced IFN-γ secretion and cell cytotoxicity. Physiological amount of sBCMA in the serum of MM patients was also measured, finding it to be around 100 ng/mL [
18]. In Lee et al., the performance of anti-BCMA CAR and anti-BCMA bispecific T cell engagers was also affected by sBCMA in a dose dependent manner effectively competing with the BCMA protein present in OPM-2 or K562/BCMA + cells [
26]. In contrast, CARTemis-1 demonstrated resistance to sBCMA up to 150 ng/mL in both in vitro and in vivo experiments (Figs.
1 and
2). Depending on the epitope recognized by the anti-BCMA CAR, sBCMA may interfere with the recognition of membrane-bound BCMA on myeloma cells and diminish anti-myeloma reactivity. One possible explanation is that the target epitope of soluble sBCMA may be inaccessible as sBCMA forms homodimeric or heterodimeric complexes. Previous studies have shown that sBCMA form large complexes with APRIL (a proliferation-inducing ligand) and BAFF (B-cell activating factor), suggesting that the epitope might be concealed within these complexes, thereby not impacting the functionality of BCMA CARs [
27,
28]. Overall, CARTemis-1 has been rationally designed to enhance antitumor efficacy and we did not observe reduced antimyeloma reactivity from T cells expressing this BCMA-CAR in the presence of sBCMA.
To achieve adequate CAR-T cell doses, different protocols have been explored for ex-vivo expansion [
29]. IL-2 is the most widely used cytokine for T-cell activation and expansion; however, several studies have explored other combinations in anti-CD19 CAR-T cells [
30‐
33]. Limited information on the comparison between different expansion protocols has been described within anti-BCMA CAR products. These cytokines play pivotal roles in T-cell function. IL-2 induces TCR-activated T-cell proliferation, but excessive levels can lead to antigen-induced cell death (AICD) [
34]. Notably, IL-2 is a key contributor to Treg proliferation. This population has recently become relevant in the field, as an increased proportion of CAR-Tregs (> 5% CAR-Tregs) in infusion products has been correlated with a lower probability of response [
35,
36]. IL-7, which is vital for T-cell homeostasis, maintains naïve and memory-resting T-cells, enhancing survival through the PI3KCLO-AKT pathway and increasing the expression of antiapoptotic factors [
37,
38]. IL-15 supports memory T-cell proliferation and survival. IL-15 stimulation reduces mTORC1 activity, shifts T-cell metabolism to favor long-lived memory T-cells, increases less-differentiated CAR-T cells, enhances cell survival, and reduces exhaustion marker expression, which is correlated with heightened antitumor responses [
39,
40]. However, while IL-15 stimulation has been reported to increase Treg accumulation in vitro [
41], the concentration used in that study was up to 10 times higher than what we used for CARTemis-1 expansion. As a result, the proportion of Tregs in our CARTemis-1 product was minimal, averaging 1.44%. On the contrary, IL-2 is critical for Treg survival and functions as a key growth factor for these cells due to their high expression of the high-affinity IL-2 receptor CD25 [
42‐
44]. Therefore, the combination of IL-7 and IL-15 is less likely to increase Treg percentages compared to IL-2. Taken altogether, these studies of CD19 CAR-T cells, are in accordance with the preclinical and clinical findings on our BCMA CAR product, in which the combination of IL-7 and IL-15 during the manufacturing process increased T-cell expansion and allowed the generation of less-differentiated and less exhausted CARTemis-1 cell products, maintaining hallmarks of T cell activation.
In addition to stimulating cytokines, cell culture period is a critical aspect of CAR-T cell therapy. Patients included in CAR-T cell therapy clinical trials are generally heavily pretreated [
29]. Short cell culture periods (3–5 days) have been described [
45‐
47]. However, this short ex vivo expansion may not be suitable for heavily pretreated patients with reduced and dysfunctional T cells [
48]. In the present study, we characterized CARTemis-1 cells during the manufacturing process to define the optimal cell culture period. Although CAR-T cell dose was achieved on day 6 postactivation for the three GMP-validation processes, we observed CARTemis-1 cells exhibited increased overexpression of coinhibitory markers early post-transduction (day 4 post-transduction = day 6 postactivation) and that this exhausted phenotype decreased at days 8–10 postactivation, both at preclinical and clinical scales (Fig.
4f). Some recent studies have reported the feasibility of generating CAR-T cells in 22–36 h (FasT CAR-T) [
49]; however, we demonstrated that early after T cell transduction, coinhibitory marker levels are significantly increased and therefore, at that time, the fitness of the product may not be optimal, particularly considering that some studies have reported that increased expression of coinhibitory markers in the infusion product is associated with worse outcomes [
50‐
53]. In the present study, we propose from day 8 on as the optimal release time point for obtaining the best-fit CARTemis-1 product (days 8–10). In this sense, in addition to obtaining a sufficient CAR-T cell dose for infusion, monitoring the immunophenotype of the product during the manufacturing process may also be an important criterion to determine the optimal product.
Several limitations exist when generating patient-derived CAR-T cells for myeloma patients. First, obtaining sufficient T cells may be challenging because patients are often lymphopenic and have dysfunctional T cells [
54]. Second, the characteristics of the leukapheresis material also have an impact on CAR-T cell fitness [
55]. Finally, the number of prior treatments [
56], patient age [
57,
58], and the disease itself [
32,
59] have been associated with limitations in the number and quality of CAR-T products. To overcome these limitations, we propose the possibility of generating CARTemis-1 cells from the small proportion of MM patients who undergo allogeneic stem cell transplantation (STC) and relapse after transplantation. However, this approach may present some drawbacks, such as the potential risk of inducing GvHD. Previous studies have analyzed the incidence of GvHD in patients receiving CAR-T cell therapy postallogeneic stem cell transplantation and found no increase in the incidence of GvHD [
55,
60‐
63]. Despite the absence of clinical results with CARTemis-1 at this point, our mouse models suggest that CARTemis-1 cells would not increase GvHD incidence compared to PBMCs, despite the high levels of activation. In summary, CARTemis-1 might also be used for the treatment of the small subset of high-risk MM patients undergoing allo-SCT, which are clearly underrepresented in clinical trials.
Recently, the FDA reported the detection of T cell malignancies, including CAR-T-cell-positive lymphoma, in patients receiving commercially available CAR-T cell products [
64]. Although the overall benefits of these products outweigh the potential risks, several international scientific experts have appointed the requirement for long-term follow-up in post monitorization studies to unravel the possible causal link between CAR-T therapy and these rare cases of T-cell malignancy [
65]. In this sense, we have also incorporated a safety-gene consisting of the truncated epidermal growth factor receptor (EGFRt), which allows both the in vivo monitoring of CARTemis-1 using anti-EGFR antibodies and the elimination of CAR-T cells in vivo with cetuximab, an anti-EGFR antibody approved for use in the clinical setting.
Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.